Generation of a human IgM monoclonal antibody ... - Springer Link

10 downloads 71 Views 578KB Size Report
These results suggest that this human IgM monoclonal antibody against HLA-class II could be considered as a potential agent in the treatment of several ...
Cancer Immunol Immunother (2009) 58:351–360 DOI 10.1007/s00262-008-0558-6

O R I G I N A L A R T I CL E

Generation of a human IgM monoclonal antibody directed against HLA class II molecules: a potential agent in the treatment of haematological malignancies Belén Díaz · Irene Sanjuan · Francisco Gambón · Carmen Loureiro · Susana Magadán · África González–Fernández

Received: 9 October 2007 / Accepted: 27 June 2008 / Published online: 2 August 2008 © Springer-Verlag 2008

Abstract Major histocompatibility complex (MHC) class II molecules have been considered as a good target molecule for use in immunotherapy, because of the high expression in some lymphoma and leukaemia cells and, also, because of their restricted expression on human cells (monocytes, dendritic, B lymphocytes, thymic epithelial cells, and some cytokine-activated cells, such as T lymphocytes). We have obtained a human IgM monoclonal antibody directed against human leukocyte antigen (HLA) class II molecules, using transgenic mice carrying human Ig genes. The antibody BH1 (IgM/ isotype) recognises HLA-class II on the surface of tumour cells from patients suVering from haematological malignancies, such as chronic and acute lymphocytic leukaemias, non-Hodgkin lymphomas and myeloid leukaemias. Interestingly,

Belén Díaz, Irene Sanjuan and Susana Magadán share authorship; Francisco Gambón and África González–Fernández share leadership. B. Díaz (&) · Á. González–Fernández Immunology Unit, Universidad de Vigo, EdiWcio de Ciencias Experimentales, Campus Lagoas Marcosende, s/n, 36310 Vigo, Pontevedra, Spain e-mail: [email protected] I. Sanjuan · F. Gambón Immunology Unit, Hospital Meixoeiro, Complejo Hospitalario Universitario de Vigo, 36300 Vigo, Pontevedra, Spain C. Loureiro Hematology Unit, Hospital Meixoeiro, Complejo Hospitalario Universitario de Vigo, 36300 Vigo, Pontevedra, Spain S. Magadán Instituto Superior de Saude do Alto Ave (ISAVE), Quinta de Matos, Geraz do Minho, 4830-31 PVL, Portugal

functional studies revealed that BH1 mAb recognises and kills very eYciently tumour cells from several leukaemia patients in the presence of human serum as a source of complement. These results suggest that this human IgM monoclonal antibody against HLA-class II could be considered as a potential agent in the treatment of several malignancies. Keywords HLA class II · Human monoclonal antibodies · Immunotherapy · Leukaemia

Introduction Although murine mAbs are relatively easy to produce [19], and some chimeric mouse–human or humanized mouse– human anti-human mAbs are now available [1, 5], there are severe restrictions on their therapeutic use in humans, due to their immunogenicity [9, 13] and the resulting reduction in their eYcacy and safety. These limitations could be overcome by the use of fully human mAbs [4, 29, 37], which would allow their repeated administration without immunogenic and/or allergic response. The development of transgenic mouse strains, engineered with unrearranged human Ig genes, appears to be a good solution for the generation of speciWc human mAbs [8, 21, 28]. The human class II molecules (DR, DP, DQ) of the major histocompatibility complex (MHC) in humans are, under normal conditions, selectively expressed on immune cells, including B lymphocytes, activated T lymphocytes, monocytes and dendritic cells. Moreover, the class II molecules of the human leukocyte antigen system (HLA) are expressed on many human B and myeloid leukaemias, as well as on B cell lymphomas, at high surface densities [23].

123

352

Recently, some murine anti-HLA class II mAbs have been modiWed by molecular biology techniques to develop either humanized or chimeric antibodies. Thus, Hu1D10 (humanized) and CHLym-1 (chimeric human–mouse) mAbs antibodies are currently being evaluated for the treatment of human B cell lymphomas, although the clinical application of antibodies could still be restricted by their potential immunogenicity in humans. Indeed, it has been shown that Hu1D10 could cause severe type-1 hypersensitivity reactions and clinical symptoms including increased heart rate, dyspnoea, vomiting and urticaria [33]. More recently, two fully human anti-HLA class II antibodies termed 1D09C3 [26] and HD8 [35] have been generated by the screening of a human combinatorial antibody library, and by the use of a transchromosome mouse that bears human immunoglobulin genes, respectively. Both antibodies have an intrinsic tumoricidal activity in vitro in cell lines, therefore they are promising immunotherapeutic agents. Here, we report the generation of a new human monoclonal antibody called BH1. This human mAb, a human IgM/, was obtained from a transgenic BAB ,  mouse that carries human transgenes for  heavy, and  and  light chains, in an inactivated endogenous IgH and Ig background [2, 28]. This mAb speciWcally recognises human HLA-class II molecules on the surface of human monocytes, B lymphocytes, tumour B cell lines (Hmy, Raji and Daudi) and tumour cells from patients suVering from haematological malignancies. It is demonstrated that BH1 shows tumoricidal eVect on human leukaemia cells from patients suVering from several disorders, killing those cells in the presence of complement. Competitive assays using BH1 and other anti-HLA class II mAbs (Q2/70 and CHLym-1) indicate that both BH1 and Q2/70 mAbs recognise the same HLA class II antigenic epitope, not shared by the chimeric CHLym-1 mAb. Although most of the mAbs being used as therapeutic agents in haematological malignancies are of IgG isotype [1, 11, 18], the value of IgM mAbs in human therapy should also be considered, due to their high eYcacy in complement activation and lower level of entry into normal tissues. The results of this study support the development of a promising potential therapeutic use for human IgM mAbs, and BH1 can be a suitable in vivo candidate in antibody therapy for the removal or possible eradication of tumour cells.

Materials and methods Experiments reported here were approved by the Ethical Committee of the University of Vigo and the Xunta de

123

Cancer Immunol Immunother (2009) 58:351–360

Galicia (Spain), in accordance with the Declaration of Helsinki. Transgenic mice BAB ,  mice [22, 28] were provided by Dr Marianne Brüggeman (Babraham Institute, Cambridge, England) and maintained under Wltered air in-Xow cabinets and fed with irradiated food and autoclaved water at the Animal Facility of the University of Vigo. Cell lines The human tumour cell lines used in this study were: Hmy, Raji and Daudi (HLA class II positive B cell lines, [12, 15, 31]), Jurkat (T cell line); U937 (histiocytic lymphoma cell line); K562 (erythroleukaemia); RPMI 8266; MM1R and M144 (myeloma cells), PC3 (prostate carcinoma); Colo205 (colon carcinoma); MCF7 (breast carcinoma); and ME180 (cervical carcinoma). All cells were cultured in DMEM or RPMI 1640 medium (Gibco, Life Technologies, Grand Island, Scotland) supplemented with 10% heat inactivated foetal calf serum (FCS) (PAA, Linz, Austria), penicillin (100 U/mL) and glutamine (2 mM) (Gibco) at 37 C in a humidiWed atmosphere containing 5% CO2. Human cell samples Heparinized whole blood from healthy donors was used for the analysis of several blood cell populations (lymphocytes, granulocytes, erythrocytes, monocytes and platelets). Blood was diluted with PBS 1£, and human peripheral blood mononuclear cells (huPBMC) were isolated by density gradient centrifugation over Ficoll-PaqueTM solution (Amersham Pharmacia Biotech AB, Uppsala, Sweden). In activation assays, huPBMC were activated for 48 h in the presence or absence of 10 g/mL of PHA (lectin from Phaseolus vulgaris) (Sigma). For the analysis of granulocytes, distillated water was added to whole blood to eliminate erythrocytes. Platelets were obtained by centrifugation of diluted blood at 40g for 5 min, and supernatant was collected. Erythrocytes were obtained by centrifugation of diluted blood at 231g for 5 min, and cellular pellet was obtained. The Immunology and Haematology Units of the Hospital Meixoeiro of Vigo provided peripheral blood and/ or bone marrow from leukaemia patients. Bone marrow cells were obtained by aspiration for diagnostic purposes, and mononuclear cells were isolated as described above. Cellular suspensions were also obtained from tumour lymph nodes (lymphomas), or from normal lymph nodes from patients without tumour pathology, who were undergoing surgery.

Cancer Immunol Immunother (2009) 58:351–360

Immunization, cell fusion and screening Transgenic mice were immunized twice (3 weeks interval) with 5–10 £ 106 viable Hmy cells by intraperitoneal injection in the presence of complete and incomplete Freund Adjuvant (CFA, Gibco Laboratories, Detroit, Michigan, USA), respectively. Mouse sera were tested 2 weeks later for the production of human IgM antibodies by ELISA and by indirect immunoXuorescence (see below). Three or Wve days before fusion, mice were intravenously boosted with 5 £ 106 viable Hmy cells. The spleen was removed and splenocytes were fused to NSO mouse myeloma cells using PEG 1500 (Sigma). The hybridomas generated were screened for secretion of HuIgM antibody by ELISA [22] or by Xow cytometry against target cells. Flow cytometry Flow cytometry analysis was carried out using diVerent target cells. The 3 £ 105 cells were incubated at 4 C for 30 min with medium alone, neat or concentrated hybridoma supernatants and washed twice with PBS. Cells were stained with FITC-rabbit anti-HuIgM Abs (Dako) at 4 C for 20 min. The stained cells were washed twice with PBS and the cellular Xuorescence was measured using a XL-MCL Flow Cytometer (Coulter Electronic, Hialeah, FL). The isotype of the secreted BH1 mAb was determined using FITCrabbit anti-human , anti-human  light chain (Dako) or FITC goat anti-mouse  light chain (Caltag, Burlingame, CA) For two-colour staining studies, cells were incubated with PE or FITC-conjugated mouse anti-HLA-DR mAb (Immunotech, Beckman Culter, Inc. Fullerton, CA, USA), washed twice with PBS and incubated with BH1 followed by FITC or PE-rabbit anti-HuIgM Ab as second stage reagent, respectively. In some cases, cells were also studied using a mouse anti-HLA class II Q2/70 mAb (kindly provided by Dr Charo de Pablo, Clínica Puerta de Hierro, Madrid) [32], and human BH1 mAb, followed by incubation with the corresponding PE, FITC- anti-mouse Igs or anti-human IgM, respectively. The analysis of diVerent cell populations was undertaken using forward and size scatter with the mAbs directed against the speciWc surface markers CD7, CD20, CD14, CD13, CD41 (Immunotech, Beckman Culter, CA) for T and B cells, monocytes, granulocytes, and platelets, respectively, and Ery-1 (CBG, Spain) for erythrocytes. A human IgM mAb (hAIM-29) against human CD69 molecule [24] was used as isotype control. Competitive assay A competitive cell binding assay was carried out to assess the reactivity and speciWcity of BH1 against HLA class II molecules. Raji and Hmy cells were incubated with a

353

constant amount of BH1 (60 ng) and varying quantities of supernatants containing mouse Q2/70 [32] or chimeric mouse–human IgG CHLym-1 [17] (kindly provided by Dr Charo de Pablo and Dr Alan Epstein, respectively), at 4 C for 45 min. After washing, cells were incubated with FITC or PE-rabbit anti-HuIgM Abs as second-stage reagents. Cell staining was analysed using a XL-MCL Flow Cytometer. Western blot analysis and immunoprecipitation In 1 mL of lysis buVer (250 mM NaCl, 25 mM Tris-HCl pH 7.5, 5 mM EDTA pH 8.0 and 0.1–1% NP-40), 107 cells were incubated at 4 C for 30 min, in the presence of protease inhibitors (Complete-mini, Roche Diagnostics, USA). Samples were cleared by centrifugation at 11g for 10 min. The amount of protein in the supernatants was measured by the Bradford Colorimetric method. For immunoblots, 10 g of lysates were separated on 12% SDS-PAGE. Proteins were transferred using a Trans-blot Semi-dry (BioRad Laboratories, Cambridge, MA) onto PVDF membrane (Hybond-P, Amersham Pharmacia Biotech, Buckinghamshire, England). The membranes were incubated with neat or concentrated hybridoma supernatant containing BH1 mAb, followed by incubation with alkaline phosphatase rabbit anti-human IgM Abs (Dako). 5-Bromo-4-chloro-3indolyl phosphate dipotassium/nitrotetrazolium blue chloride (BCIP/NBT, Sigma) was used as substrate to visualise speciWc protein. For immunoprecipitation, the mouse antiHLA-class II Q2/70 mAb was coupled to Protein G (Pierce, Rockford, IL) according to the manufacturer’s instructions. Protein G-Q2/70 was incubated with Hmy and Jurkat lysates, washed several times and the resulting samples were applied on 12% SDS-PAGE, transferred onto PVDF membrane and incubated with BH1 as described above. Complement-dependent cytotoxicity (CDC) assay The ability of BH1 to kill diVerent cells was determined by CDC assay and analysed by Xow cytometry. Leukaemia cells from patients were incubated for 30 min at 4 C with medium alone (as negative control), BA5 as isotype control (a human IgM mAb produced in our laboratory that recognises B cells isolated from normal huPBMC and some tumoral B cells) [22, 34], BH1, or with antiHLA-class II CHLym-1 and Q2/70 mAbs. Cells were washed twice and incubated at 37 C for 30 min with rabbit complement (diluted 1:2 in medium) (Dade Behring, Marhurg, Germany), human complement (Sigma), or with human serum as a source of complement. Cell viability was tested by adding propidium iodide (PI) (Sigma) and death cells were analysed by Xow cytometry for PI staining.

123

354

Cancer Immunol Immunother (2009) 58:351–360

Results A human monoclonal antibody (BH1) recognises HLA class II molecules We have previously shown that high aYnity human mAbs directed against human antigens can be obtained from transgenic BAB and BAB, mice [22, 24, 34]. Transgenic BAB, mice were immunized with human tumour B cells in order to obtain monoclonal antibodies directed against human B cells, which could be used in human therapy against B cell leukaemias. Spleen cells from transgenic mice were fused eYciently with the NSO myeloma cells, allowing the isolation of hybridomas secreting speciWc human IgM (HuIgM) directed against B cells. One of these hybridomas, which secretes BH1, a human monoclonal IgM/, was used to perform additional studies. The human mAb BH1 speciWcally recognises human B lymphocytes and monocytes (Table 1), but not T cells, granulocytes, erythrocytes or platelets from peripheral blood. In addition, it does not stain normal bone marrow cells or T cells from non-tumour lymphoid nodes (Table 1). Moreover, BH1 only stained the B cell lines Hmy, Daudi and Raji. Other tumour cells tested of diVerent origin (Jurkat, U937, K562, RPMI 8266, MM1R, M144, PC3,

Colo205, MCF7 and ME180) were not recognised by the BH1 mAb (Table 1). Several methods were used to characterise the antigen recognised by BH1. First, we analysed whether it could be expressed on human PHA-activated PBMC. The analysis of the staining showed that the antigen recognised by BH1 mAb was expressed in human CD7+ lymphocytes only after activation (Fig. 1a), indicating that it recognises a marker expressed on activated T lymphocytes. As a positive control of cell activation, the staining of HLA class II with a commercial anti-HLA-DR mAb and Q2/70, a diVerent mouse anti-HLA class II mAb, were also included, and interestingly, the staining showed a linear pattern in both cases, which strongly suggests a co-recognition by both monoclonal antibodies (BH1 and anti-HLA class II) of the same molecule (Fig. 1a). The same double immunoXuorescence analysis was carried out on Hmy, a HLA class II positive B cell line (Fig. 1b), and a perfect correlation in the staining with both Abs was also observed. In contrast, the anti-CD20 mAb, which also recognises Hmy cells, did not show a pattern of co-staining with the commercial anti-HLA-DR mAb (Fig. 1b). The conWrmation that BH1 recognises HLA class II molecules was undertaken by a competitive binding assay using BH1 and other anti-HLA-class II molecules mAbs.

Table 1 Recognition of normal and tumour human cells by human BH1 mAb %a

 MFIb

Normal cells

HMY

98.6 § 1.5

831.1 § 43.4

DAUDI

97.2 § 2.4

67.5 § 1.5

RAJI

Tumour cell lines Haematopoietic

%a

 MFIb

Lymphocytes B

97.4 § 0.1

75.9 § 8.7

Lymphocytes T

0.3 § 0.4

0.9 § 1.1

88.4 § 3.5

39.3 § 6.1

Peripheral blood

99.9 § 0.7

501.4 § 9.4

JURKAT

0.7 § 0.1

1.3 § 2.7

Granulocytes

1.3 § 1.5

0.1 § 0.2

U937

0.7 § 0.3

2.5 § 2.8

Erytrocytes

0.0 § 0.0

0.0 § 0.0

1.9 § 1.5

5.9 § 0.6

Platelets

0.0 § 0.0

0.0 § 0.0

18.7 § 1.3

6.5 § 0.3

K562 RPMI8266 MM1R

2.3 § 0.3

0.5 § 0.1

M144

17.1 § 0.5

8.3 § 0.2

(n = 3)

(n = 3)

(n = 4)

(n = 4)

Lymph nodesc

Non-haematopoietic PC3

1.6 § 0.1

4.3 § 0.3

Lymphocytes B

92.6 § 5.5

86.2 § 6.9

Lymphocytes T

1.1 § 0.8

2.1 § 0.7

Colo205

0.9 § 0.2

0.6 § 0.1

MCF7

0.8 § 0.3

0.4 § 0.1

ME180

0.6 § 0.2

0.3 § 0.1

(n = 3) a

Monocytes

(n = 3)

Bone marrow mononuclear cellsc

(n = 3)

(n = 3)

6.6 § 5.3 (n = 10)

2.6 § 2.5 (n = 10)

Percentage (%) of cells recognised by BH1. The results are expressed as Mean § sd obtained from n independent experiments Mean Xuorescence intensity (MFI) calculated as: MFI(BH1 + secondary antibody) ¡ MFI(secondary antibody). It is expressed as the Mean § SD obtained from n independent experiments c Non-tumour samples obtained from lymph nodes or bone marrow b

123

Cancer Immunol Immunother (2009) 58:351–360

355

Fig. 1 Two-colour staining using PHA-activated PBMC as target cells (a) or the Hmy cell line (b). Cells were incubated with directly conjugated (PE or FITC) anti-HLA-DR, anti-CD7, and anti-CD20 mAbs, and with mouse Q2/70 or BH1 mAbs (followed by FITC-rabbit anti-mouse IgG and PE-rabbit anti-human IgM antibodies, respectively)

Hmy or Raji cells were incubated in the presence of a constant amount of BH1 and varying amounts of Q2/70 or CHLym-1. As shown in Fig. 2a, while Q2/70 competes with BH1, this does not happen for CHLym-1 (Fig. 2b). Increasing amounts of Q2/70 decrease the staining of cells to the BH1 mAb, indicating that both antibodies recognise the same antigenic epitope. Furthermore, Western blot analysis of cell lysates showed that BH1 binds a protein of 28 KDa, which corresponds to the expected molecular weight for the HLA class II-beta chain, on Hmy and Daudi (both B cell lines), but not on Jurkat cells (Fig. 2c). BH1 was able to stain the same band when the HLA class II antigen was immunoprecipitated with Protein G-Q2/70 mAb (Fig. 2c). All these results conWrm that BH1 recognises HLA class II antigen. BH1 mAb recognises and kills cells of human haematological disorders Malignant cells from the peripheral blood, bone marrow or lymph nodes of 80 patients suVering from diVerent haematological malignancies were tested to demonstrate the speciWcity of BH1 against malignant cells. Table 2 shows the summary of the patients analysed and the level of recognition of BH1 to tumour cells. Over 86% of the chronic lymphocytic leukaemias (CLL), 30 out of 35, were recognised by the mAb BH1. Tumour cells from four patients included in the study with acute lymphocytic leukaemia (ALL) were also stained. Moreover, 60% of the total samples analysed including acute- (AML) and chronic myeloid leukaemia

(CML) were stained by BH1 mAb, and 80% of the nonHodgkin lymphoma tested, 12 out of 15 cases. In all cases of positive recognition, tumour cells were HLA-DR+, which was further conWrmed by staining with commercial anti-HLA-DR mAbs (data not shown). Myeloma (M) cells were not recognised by the BH1 mAb in any of the 3 cases tested, showing similar results to those with the myeloma cell lines (Table 1). However, some myeloma cell lines were stained in low expression with the commercial antiHLA-DR+, but not with our mAb BH1 (data not shown). Complement-dependent cytotoxicity (CDC) assays were performed with samples from patients with lymphoproliferative disorders in the presence of BH1 mAb, and rabbit or human complement. The BH1 mAb recognised 15 out of 18 of the leukaemia samples tested. In those cases of positive staining, BH1 was able to induce cell death in the presence of rabbit complement in all cases (percentage of death was variable ranging from 48 to 99%, depending on the tumour DR expression). Interestingly, BH1 also killed tumour cells in the presence of human serum as a source of complement in a similar way (an example is shown in Fig. 3a). To conWrm that death cells correspond to tumour HLA-DR+cells, leukaemia cells were stained with antiCD20 and anti-HLA-DR before and after incubation with BH1 and complement. Figure 3b shows how BH1 mAb is able to kill almost all tumour CD20+ HLA-DR+ cells of a patient suVering from CLL, in the presence of human serum, in only 30 min. These results show that the BH1 is able to activate human complement and induce a very eYcient killing of human leukaemia cells.

123

356

a

COMPETITIVE ASSAY Hmy

Raji Negative

N° cells

Fig. 2 Competitive assay. Hmy and Raji cells were incubated with a low amount of BH1 mAb and increasing quantities of mouse Q2/70 (a) or chimeric CHLym-1 mAbs (b). BH1 mAb was detected with PE or FITC conjugated anti-human IgM Abs. c BH1 mAb recognises a protein of 28 KDa on Hmy and Daudi lysates (both B cell lines), but not on Jurkat (T cell line) in Western blot analysis (left blot). Immunoprecipitation of HLAclass II antigen using the protein G-Q2/70 mAb, stained with BH1 mAb followed by alkaline phosphatase rabbit anti-human IgM Abs (right blot)

Cancer Immunol Immunother (2009) 58:351–360

BH1 BH1 + Q2/70 (15 ng) BH1 + Q2/70 (35 ng) BH1 + Q2/70 (70 ng)

BH1 + anti-huIgM-PE

b

Hmy

Raji

Negative BH1

N° cells

BH1 + CHLym (15 ng) BH1 + CHLym (35 ng) BH1 + CHLym (70 ng)

BH1 + anti-huIgM-FITC

c

IMMUNOPRECIPITATION

CELL LYSATES Daudi Jurkat Hmy

Jurkat

28 KDa

28 KDa

A dose–response curve was produced comparing BH1 with the other HLA class II mAbs (mouse Q2/70 and the chimeric human–mouse CHLym-1), in their ability to induce cell death in the presence of human complement. Samples from ten patients suVering from CLL were included in this study, using limiting concentrations (ranging from 1 to 2 £ 10¡4 g/mL) of antibodies (Fig. 4). Commercial human complement was used instead of serum, to try to avoid the diVerences that could be attributed to complement activity in human serum. The results showed a large variation in the percentage of cell lysis between samples, independently of their HLA class II expression levels. Thus, patients were classiWed into two groups, according to their CDC susceptibility: CDC sensitive (Fig. 4a) and CDC resistant (Fig. 4b). For CDC sensitive samples, the mean of cell lysis was over 50% (using 1 or 0.2 g/mL of antibody), while CDC resis-

123

Hmy

tant samples showed a mean of 6% of cell lysis. Interestingly, all CDC-resistant samples were from patients being treated by chemotherapy at the time of study, although there were still tumoral cells (HLA-DR+) in peripheral blood. Despite these diVerences between samples, the percentage of cell lysis induced by BH1, Q2/70 and CHLym-1 was very similar at all concentrations tested (Fig. 4). As expected, cell death was strongly dependent on antibody concentration, and lower Ab concentrations, induced lower levels of death.

Discussion Antibodies and immunoconjugates are gaining a signiWcant and expanding role in cancer therapy. The HLA-class II

c

60/80 Total

b

19.8 (n = 1) 64.1 § 8.8 (n = 8) 12/15

0/3 Myeloma (ML)

Non-Hodgkin lymphoma (NHL)

7/10 Acute myeloid leukaemia (AML)

34.2 (n = 1)

60.5 § 1.6 (n = 3)

5.3 § 2.6 (n = 3)

21.1 § 2.2 (n = 2) 55 § 14.8 (n = 5)

7/13 Chronic myeloid leukaemia (CML)

32.9 § 26.6 (n = 3)

55.4 § 26.6 (n = 4)

67.7 § 15.1 (n = 4) 4/4 Acute lymphocytic leukaemia (ALL)

Bone marrow

84.3 § 5.7 (n = 5) 74.9 § 1.7 (n = 25) 30/35 Chronic lymphocytic leukaemia (CLL)

a

7.4 § 10.8 (n = 3)

3.9 § 2.9 (n = 2) 14.3 § 7.6 (n = 5)

23.5 § 51.1 (n = 3)

18.7 § 20.8 (n = 4)

11.1 § 3.5 (n = 4)

39.8 § 41.6 (n = 5) 32.7 § 32.9 (n = 25)

Bone marrow PBL PBL

Lymph nodes

MFIc % of cells recognisedb CR/na

Table 2 Recognition of tumour cells by human BH1 mAb from 80 patients suVering from diVerent haematological malignancies

CR/n, number of cases recognised by BH1/number of cases tested (n) Percentage (%) of cells recognised by BH1. The result is expressed as the Mean § SD Mean Xuorescence Intensity (MFI) obtained as MFI(BH1 + secondary antibody) ¡ MFI(secondary antibody). The result is expressed as the Mean § SD

32.1 § 54.9 (n = 8)

molecule is an emerging therapeutic target in haematological malignancies and several anti-HLA class II mAbs have been developed, such as CHLym-1 (chimeric) [11] and Hu1D10 (Apolizumab, a humanized mAb) [13, 25], although some adverse eVects have been reported for the humanized mAb Hu1D10 [33]. BAB, transgenic mice were used in this study to generate BH1, a human IgM mAb. Using several approaches, we demonstrate that BH1 is directed against HLA-class II molecules. The competition assay indicates that BH1 recognises a shared epitope with the Q2/70 mAb, which has been shown to interact with the beta chain of both HLA class II DR and DQ molecules [27]. We also show that BH1 recognises and kills primary leukaemia target cells in the presence of complement, suggesting that this antibody could provide an eVective therapy in some tumour haematological processes (mainly lymphocytic, also some myeloid leukaemias). Very recently, two additional human IgG anti-HLA-DR antibodies (1D09C3 and HD8) have also been generated [6, 7, 35], which show tumoricidal activity, although their eYcacy in human patients has still to be conWrmed. It is reasonable to think that no immune responses would be generated against either of these human antibodies or against our BH1 mAb, bearing in mind that all are fully human. Human immunotherapy generally uses mouse, chimeric or humanized antibodies, mainly of the IgG isotype [16, 20, 36] due to their speciWcity and higher level of tumour penetration, although other isotypes (IgM and IgA) have also been considered [3, 10]. The eYcacy of these anti-tumour antibodies usually depends on their capacity of activating immunological eVector mechanisms, such as complement activation and, sometimes, they are not very eVective because human cells produce speciWc complement-inhibitory factors, which confer resistance to their own serum complement. To avoid this resistance, some mAbs must be used in combination. For example, the anti-CD20 mAb is used together with antibodies to block some of the complement-inhibitory factors (e.g. CD55 or CD59 molecules) [14, 36, 38]. This increases the eYcacy of the therapy, but also its cost and the possibility of secondary eVects. In this sense, BH1 could have some advantages compared to IgG because the IgM isotype is a very good complement activator. Furthermore, no allotypic diVerences have been described in the IgM isotype, in contrast to IgG antibodies [30], although its inXuence on antibody immunogenicity could be very low. Additionally, due to the high molecular weight of IgM, a minor level of entry into normal tissues should be expected, probably with lower secondary eVects. If necessary, the isotype of the BH1 mAb could always be changed to IgG.

56.2 § 27.5 (n = 3)

357

Lymph nodes

Cancer Immunol Immunother (2009) 58:351–360

123

358

a

RABBIT COMPLEMENT MEDIUM

N° cells

89%

HUMAN COMPLEMENT

92%

8% PI

4% FS

BA5

85%

83% N° cells

Fig. 3 a Target cells were incubated with rabbit (left) or human complement (right) in the presence of medium, BA5 (an isotypic Ab control) or BH1 mAb. The viability of the cells was tested by the incorporation of propidium iodide (PI) by FACS analysis. b Leukaemia cells from a patient suVering from CLL were analysed by Xow cytometry with anti-CD20 PE and antiHLA DR FITC mAbs, before (left) and after (right) incubation with BH1 mAb and human serum as a source of complement

Cancer Immunol Immunother (2009) 58:351–360

9% PI

5% FS

N° cells

BH1

17%

30%

80% PI

66% FS

Leukaemia cells (CLL)

CLL+ BH1 + HUMAN C´

CD20 - PE

b

DR - FITC

The present study shows that BH1 is able to kill tumour cells not only in the presence of rabbit complement, but also with human complement. Its ability to activate CDC was almost identical to other anti- HLA class II mAbs (Q2/ 70 and CH Lym-1), all of them showing a dose-dependent eVect (Fig. 4). However, a large variation in killing has been found. In the presence of BH1 and rabbit complement, where 15 patient samples were analysed, cell lysis ranged from 49% to nearly 100%, and death was found in all samples tested. Yet, for human complement, two diVerent groups can be distinguished: sensitive or resistant (Fig. 4). These diVerences in CDC susceptibility could be due to several factors, such as a variable production of complement-

123

inhibitory molecules, the presence of complement-resistant cell clones in some patients (indeed, all resistant samples were from patients being treated by chemotherapy) or by other undetermined factors [14, 36, 38]. Although few samples were included in this CDC study, and many more in vitro and in vivo trials are needed, the results indicate that an early introduction of antibodies in therapy could be the best choice. This could also be true for other mAbs. It is worth pointing out that BH1 was not able to bind to all HLA-class II+ cells tested, especially some myeloma cell lines. A similar situation has been described with other anti-HLA class II mAbs, such as Hu1D10 and CHLym-1, which failed to recognise all HLA class II positive cells

Cancer Immunol Immunother (2009) 58:351–360

a

c

CDC SENSITIVE (N= 6)

CDC SENSITIVE

100 BH1

75

% Lysis

Fig. 4 Dose–response in complement-dependent cytotoxicity (CDC) assays. CLL samples of 10 patients were incubated with diVerent amounts of anti-HLA class II (BH1, Q2/70 and ChLym1) mAbs and commercial human complement. Patients were grouped according to their sensitivity to complement-mediated lysis in sensitive (a) and resistant (b). The results are indicated as the mean and standard deviations. Percentage of recognition and CDC lysis induced by BH1 (0.2 g/mL) on sensitive (c) and resistant patients (d)

359

Q2/70 LYM-1 50

25

0 1

0,2

0,04

0,008

0,0002

Patient

BH1(%)

Lysis(%)

1

78

16

2

50

41

3

61

15

4

80

97

5

80

86

6

80

63

mAb concentration [µg/mL]

b

d

CDC RESISTANT (N= 4)

CDC RESISTANT

25

Q2/70

% Lysis

Patient

BH1(%)

Lysis(%)

7

98

5

8

85

9

9

82

4

10

55

2

BH1

20

LYM-1

15 10 5 0 1

0,2

0,04

0,008

0,0002

mAb concentration [µg/mL]

tested [35]. The HLA class II is extremely polymorphic due to its highly variable -chain (more than 500 diVerent HLA-DR alleles have been identiWed) and, probably, BH1 and the other human mAbs can bind to some polymorphic variants, but not to all of them. This evidence shows that, to avoid treatment failures, it is essential to test the antibodies against tumour target cells before starting the immunotherapy process, and that a variety of anti-HLA-class II human mAbs is necessary to cover diVerent alleles. Thus, our human BH1 mAb could be a good candidate as a therapeutic agent for the treatment of diVerent HLA-class II positive leukaemias and other malignancies. Acknowledgments We would like to thank Dr Marianne Brüggemann for supplying the transgenic mice used in this work; Angel Torreiro for assistance with maintenance of the mice; Charo de Pablo (Clínica Puerta de Hierro, Madrid) and Professor Alan Epstein (University of Southern California, USA) for providing antibodies Q2/70 and CHLym-1, respectively; and Darío Alves, Cristian Sánchez, Daniel Pérez, Elina Garet, Silvia Lorenzo and Marisa Abad for technical help. The contributions of B. D. and I. S. include the generation and characterisation of monoclonal antibodies, analysis of peripheral blood cells and complement lysis. S. M. performed most of the Xow cytometry studies in diVerent populations and supervised the complement lysis and analysis in leukaemia patients. F. G. and A. G. supervised the work undertaken by B. D., I. S. and S. M. in the Hospital and in the University, respectively. This study was supported by the Xunta de Galicia, the Instituto de Salud Carlos III (Red del FIS G03/136) and the Ministerio de Educación y Ciencia (Nanobiomed, ConsoliderIngenio2010).

ConXict of interest statement Wnancial interests.

The authors declare no competing

References 1. Adams GP, Weiner LM (2005) Monoclonal antibody therapy of cancer. Nat Biotechnol 23:1147–1157 2. Brüggemann M, Caskey HM, Teale C, Waldmann H, Williams GT, Surani MA, Neuberger MS (1989) A repertoire of monoclonal antibodies with human heavy chains from transgenic mice. Proc Natl Acad Sci 86(17):6709–6713 3. Brüggemann M, Teale C, Clark M, Bindon C, Waldmann H (1989) A matched set of rat/mouse chimeric antibodies identiWcation and biological properties of rat H chain constant regions mu, gamma 1, gamma 2a, gamma 2b, gamma 2c, epsilon, and alpha. J Immunol 142:3145–3150 4. Burgess T, Coxon A, Meyer S, Sun J, Rex K, Tsuruda T, Chen Q, Ho SY, Li L, Kaufman S, McDorman K, Cattley RC, Sun J, Elliott G, Zhang K, Feng X, Jia XC, Green L, Radinsky R, Kendall R (2006) Fully human monoclonal antibodies to hepatocyte growth factor with therapeutic potential against hepatocyte growth factor/ c-Met-dependent human tumors. Cancer Res 66:1721–1729 5. Carlo-Stella C, Di Nicola M, Turco MC, Cleris L, Lavazza C, Longoni P, Milanesi M, Magni M, Ammirante M, Leone A, Nagy Z, GioVre WR, Formelli F, Gianni AM (2006) The anti-human leukocyte antigen-DR monoclonal antibody 1D09C3 activates the mitochondrial cell death pathway and exerts a potent antitumor activity in lymphoma-bearing nonobese diabetic/severe combined immunodeWcient mice. Cancer Res 66:1799–1808 6. Carlo-Stella C, Guidetti A, Di Nicola M, Longoni P, Cleris L, Lavazza C, Milanesi M, Milani R, Carrabba M, Farina L, Formelli F,

123

360

7.

8.

9.

10.

11.

12.

13.

14.

15.

16.

17.

18.

19. 20.

21. 22.

23.

Cancer Immunol Immunother (2009) 58:351–360 Gianni AM, Corradini P (2006) CD52 antigen expressed by malignant plasma cells can be targeted by alemtuzumab in vivo in NOD/ SCID mice. Exp Hematol 34:721–727 Carlo-Stella C, Guidetti A, Di Nicola M, Lavazza C, Cleris L, Sia D, Longoni P, Milanesi M, Magni M, Nagy Z, Corradini P, Carbone A, Formelli F, Gianni AM (2007) IFN-gamma enhances the antimyeloma activity of the fully human anti-human leukocyte antigen-DR monoclonal antibody 1D09C3. Cancer Res.1 67(7):3269–3275 Davis CG, Jia XC, Feng X, Haak-Frendscho M (2004) Production of human antibodies from transgenic mice. Methods Mol Biol 248:191–200 Dechant M, Bruenke J, Valerius T (2003) HLA class II antibodies in the treatment of hematologic malignancies. Semin Oncol 30:465–475 Dechant M, Vidarsson G, Stockmeyer B, Repp R, Glennie MJ, Gramatzki M, van de Winkel JG, Valerius T (2002) Chimeric IgA antibodies against HLA class II eVectively trigger lymphoma cell killing. Blood 100:4574–4580 Denardo GL, Tobin E, Chan K, Bradt BM, Denardo SJ (2005) Direct antilymphoma eVects on human lymphoma cells of monotherapy and combination therapy with CD20 and HLA-DR antibodies and 90Y-labeled HLA-DR antibodies. Clin Cancer Res 11:7075s–7079s Epstein MA, Achong BG, Barr YM, Zajac B, Henle G, Henle W (1966) Morphological and virological investigations on cultured Burkitt tumor lymphoblasts (strain Raji). J Natl Cancer Inst 37(4):547–559 Gilliland LK, Walsh LA, Frewin MR, Wise MP, Tone M, Hale G, Kioussis D, Waldmann H (1999) Elimination of the immunogenicity of therapeutic antibodies. J Immunol 162:3663–3671 Golay J, ZaVaroni L, Vaccari T, Lazzari M, Borleri GM, Bernasconi S, Tedesco F, Rambaldi A, Introna M (2000) Biologic response of B lymphoma cells to anti-CD20 monoclonal antibody rituximab in vitro: CD55 and CD59 regulate complement-mediated cell lysis. Blood 95:3900–3908 Haan KM, Kwok WW, Longnecker R, Speck P (2000) EpsteinBarr virus entry utilizing HLA-DP or HLA-DQ as a coreceptor. J Virol 74(5):2451–2454 Hale G, Cobbold S, Novitzky N, Bunjes D, Willemze R, Prentice HG, Milligan D, MacKinnon S, Waldmann H (2001) CAMPATH1 antibodies in stem-cell transplantation. Cytotherapy 3:145–164 Hu P, Glasky MS, Yun A, Alauddin MM, Hornick JL, Khawli LA, Epstein AL (1995) A human–mouse chimeric Lym-1 monoclonal antibody with speciWcity for human lymphomas expressed in a baculovirus system. Hum Antibodies Hybridomas 6(2):57–67 Isaacs JD, Wing MG, Greenwood JD, Hazleman BL, Hale G, Waldmann H (1996) A therapeutic human IgG4 monoclonal antibody that depletes target cells in humans. Clin Exp Immunol 106:427–433 Köhler G, Milstein C (1975) Continuous cultures of fused cells secreting antibody of predeWned speciWcity. Nature 256:495–497 Liu C, DeNardo G, Tobin E, DeNardo S (2004) Antilymphoma eVects of anti-HLA-DR and CD20 monoclonal antibodies (Lym-1 and Rituximab) on human lymphoma cells. Cancer Biother Radiopharm 19:545–561 Lonberg N (2005) Human antibodies from transgenic animals. Nat Biotechnol 23:1117–1125 Magadán S, Valladares M, Suárez E, Sanjuán I, Molina A, Ayling C, Davies SL, Zou X, Williams GT, Neuberger MS, Brüggemann M, Gambón F, Díaz-Espada F, González-Fernández A (2002) Production of antigen-speciWc human monoclonal antibodies: comparison of mice carrying IgH/kappa or IgH/kappa/lambda transloci. Biotechniques 33:680 See also 682,684 Mavromatis B, Cheson BD (2003) Monoclonal antibody therapy of chronic lymphocytic leukemia. J Clin Oncol 21:1874–1881

123

24. Molina A, Valladares M, Magadán S, Sancho D, Viedma F, Sanjuán I, Gambón F, Sánchez-Madrid F, González-Fernández A (2003) The use of transgenic mice for the production of a human monoclonal antibody speciWc for human CD69 antigen. J Immunol Methods 282:147–158 25. Mone AP, Huang P, Pelicano H, Cheney CM, Green JM, Tso JY, Johnson AJ, JeVerson S, Lin TS, Byrd JC (2004) Hu1D10 induces apoptosis concurrent with activation of the AKT survival pathway in human chronic lymphocytic leukemia cells. Blood 103:1846–1854 26. Nagy ZA, Hubner B, Lohning C, Rauchenberger R, ReiVert S, Thomassen-Wolf E, Zahn S, Leyer S, Schier EM, Zahradnik A, Brunner C, Lobenwein K, Rattel B, Stanglmaier M, Hallek M, Wing M, Anderson S, Dunn M, Kretzschmar T, Tesar M (2002) Fully human, HLADR-speciWc monoclonal antibodies eYciently induce programmed death of malignant lymphoid cells. Nat Med 8:801–807 27. Flomenberg N, Knowles RW, Williams D, Horibe K, Rosenkrantz K, Dupont B (1985) T lymphocyte clones detecting novel supertypic HLA class II allospeciWcities, vol 22. Springer, Berlin, pp 270–300 28. Nicholson IC, Zou X, Popov AV, Cook GP, Corps EM, Humphries S, Ayling C, Goyenechea B, Xian J, Taussig MJ, Neuberger MS, Brüggemann M (1999) Antibody repertoires of four- and Wvefeature translocus mice carrying human immunoglobulin heavy chain and kappa and lambda light chain yeast artiWcial chromosomes. J Immunol 163:6898–6906 29. Ostendorf T, van Roeyen CR, Peterson JD, Kunter U, Eitner F, Hamad AJ, Chan G, Jia XC, Macaluso J, Gazit-Bornstein G, Keyt BA, Lichenstein HS, LaRochelle WJ, Floege J (2003) A fully human monoclonal antibody (CR002) identiWes PDGF-D as a novel mediator of mesangioproliferative glomerulonephritis. J Am Soc Nephrol 14:2237–2247 30. Oxelius VA, Aurivillius M, Carlsson AM, Musil K (1999) Serum Gm allotype development during childhood. Scand J Immunol 50:440–446 31. Rouleau M, Namikawa R, Antonenko S, Carballido-Perrig N, Roncarolo MG (1996) Antigen-speciWc cytotoxic T cells mediate human fetal pancreas allograft rejection in SCID-hu mice. J Immunol 157(12):5710–5720 32. Quaranta V, Indiveri F, Glassy MC, Ng A, Russo C, Molinaro GA, Pellegrino MA, Ferrone S (1980) Serological, functional, and immunochemical characterization of a monoclonal antibody (MoAb Q2/70) to human Ia-like antigens. Hum Immunol 1:211–223 33. Shi JD, Bullock C, Hall WC, Wescott V, Wang H, Levitt DJ, Klingbeil CK (2002) In vivo pharmacodynamic eVects of Hu1D10 (remitogen), a humanized antibody reactive against a polymorphic determinant of HLA-DR expressed on B cells. Leuk Lymphoma 43:1303–1312 34. Suárez E, Magadán S, Sanjuán I, Valladares M, Molina A, Gambón F, Díaz-Espada F, González-Fernández A (2006) Rearrangement of only one human IGHV gene is suYcient to generate a wide repertoire of antigen speciWc antibody responses in transgenic mice. Mol Immunol 43:1827–1835 35. Tawara T, Hasegawa K, Sugiura Y, Tahara T, Ishida I, Kataoka S (2007) Fully human antibody exhibits pan-human leukocyte antigen-DR recognition and high in vitro/vivo eYcacy against human leukocyte antigen-DR-positive lymphomas. Cancer Sci 98:921–928 36. Treon SP, Mitsiades C, Mitsiades N, Young G, Doss D, Schlossman R, Anderson KC (2001) Tumor cell expression of CD59 is associated with resistance to CD20 serotherapy in patients with B-cell malignancies. J Immunother 24:263–271 37. Yang XD, Jia XC, Corvalan JR, Wang P, Davis CG (2001) Development of ABX-EGF, a fully human anti-EGF receptor monoclonal antibody, for cancer therapy. Crit Rev Oncol Hematol 38:17–23 38. Ziller F, Macor P, Bulla R, Sblattero D, Marzari R, Tedesco F (2005) Controlling complement resistance in cancer by using human monoclonal antibodies that neutralize complement-regulatory proteins CD55 and CD59. Eur J Immunol 35:2175–2183